Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 29
Filtrar
1.
J Neuroinflammation ; 15(1): 323, 2018 Nov 21.
Artigo em Inglês | MEDLINE | ID: mdl-30463579

RESUMO

BACKGROUND: Traumatic brain injury (TBI) represents a major cause of disability and death worldwide with sustained neuroinflammation and autophagy dysfunction contributing to the cellular damage. Stimulator of interferon genes (STING)-induced type-I interferon (IFN) signalling is known to be essential in mounting the innate immune response against infections and cell injury in the periphery, but its role in the CNS remains unclear. We previously identified the type-I IFN pathway as a key mediator of neuroinflammation and neuronal cell death in TBI. However, the modulation of the type-I IFN and neuroinflammatory responses by STING and its contribution to autophagy and neuronal cell death after TBI has not been explored. METHODS: C57BL/6J wild-type (WT) and STING-/- mice (8-10-week-old males) were subjected to controlled cortical impact (CCI) surgery and brains analysed by QPCR, Western blot and immunohistochemical analyses at 2 h or 24 h. STING expression was also analysed by QPCR in post-mortem human brain samples. RESULTS: A significant upregulation in STING expression was identified in late trauma human brain samples that was confirmed in wild-type mice at 2 h and 24 h after CCI. This correlated with an elevated pro-inflammatory cytokine profile with increased TNF-α, IL-6, IL-1ß and type-I IFN (IFN-α and IFN-ß) levels. This expression was suppressed in the STING-/- mice with a smaller lesion volume in the knockout animals at 24 h post CCI. Wild-type mice also displayed increased levels of autophagy markers, LC3-II, p62 and LAMP2 after TBI; however, STING-/- mice showed reduced LAMP2 expression suggesting a role for STING in driving dysfunctional autophagy after TBI. CONCLUSION: Our data implicates a detrimental role for STING in mediating the TBI-induced neuroinflammatory response and autophagy dysfunction, potentially identifying a new therapeutic target for reducing cellular damage in TBI.


Assuntos
Lesões Encefálicas Traumáticas/complicações , Encéfalo/metabolismo , Encefalite/etiologia , Encefalite/metabolismo , Regulação da Expressão Gênica/genética , Proteínas de Membrana/metabolismo , Animais , Autofagia/genética , Encéfalo/patologia , Lesões Encefálicas Traumáticas/patologia , Proteínas de Ligação ao Cálcio/metabolismo , Citocinas/genética , Citocinas/metabolismo , Modelos Animais de Doenças , Lateralidade Funcional , Proteína Glial Fibrilar Ácida/metabolismo , Fator 3-alfa Nuclear de Hepatócito/metabolismo , Humanos , Proteína 2 de Membrana Associada ao Lisossomo/genética , Proteína 2 de Membrana Associada ao Lisossomo/metabolismo , Masculino , Proteínas de Membrana/genética , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Proteínas dos Microfilamentos/metabolismo , Proteínas do Tecido Nervoso/metabolismo
2.
Brain ; 140(5): 1486-1498, 2017 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-28383676

RESUMO

We fractionated frontal cortical grey matter from human Alzheimer's disease and control subjects into four biochemically defined pools that represent four distinct compartments: soluble/cytosolic, peripheral membrane/vesicular cargo, integral lipid/membranous pools and aggregated/insoluble debris. Most of the readily extractable amyloid-ß remains associated with a lipid/membranous compartment. There is an exchange of amyloid-ß between the biochemical pools that was lost for the amyloid-ß42 species in Alzheimer's disease, consistent with the peptide being irreversibly trapped in extracellular deposits. The quantitative amyloid-ß data, combined with magnetic resonance imaging volumetric analysis of the amount of cortical grey matter in brain, allowed us to estimate the total mass of amyloid-ß in Alzheimer's disease (6.5 mg) and control (1.7 mg) brains. The threshold positron emission tomography standard uptake value ratio of 1.4 equates to 5.0 µg amyloid-ß/g of grey matter and the mean Alzheimer's disease dementia standard uptake value ratio level of 2.3 equates to 11.20 µg amyloid-ß/g of grey matter. It takes 19 years to accumulate amyloid from the threshold positron emission tomography standard uptake value ratio to the mean value observed for Alzheimer's disease dementia. This accumulation time window combined with the difference of 4.8 mg of amyloid-ß between Alzheimer's disease and control brain allows for a first approximation of amyloid-ß accumulation of 28 ng/h. This equates to an estimated 2-5% of the total amyloid-ß production being deposited as insoluble plaques. Understanding these rates of amyloid-ß accumulation allows for a more quantitative approach in targeting the failure of amyloid-ß clearance in sporadic Alzheimer's disease.


Assuntos
Doença de Alzheimer/metabolismo , Peptídeos beta-Amiloides/metabolismo , Lobo Frontal/metabolismo , Substância Cinzenta/metabolismo , Placa Amiloide/metabolismo , Doença de Alzheimer/patologia , Estudos de Casos e Controles , Lobo Frontal/patologia , Substância Cinzenta/patologia , Humanos , Imageamento por Ressonância Magnética , Neuroimagem , Tomografia por Emissão de Pósitrons , Fatores de Tempo
3.
Glia ; 64(9): 1590-604, 2016 09.
Artigo em Inglês | MEDLINE | ID: mdl-27404846

RESUMO

Type-1 interferons (IFNs) are pleiotropic cytokines with a critical role in the initiation and regulation of the pro-inflammatory response. However, the contribution of the type-1 IFNs to CNS disorders, specifically chronic neuropathologies such as Parkinson's disease is still unknown. Here, we report increased type-1 IFN signaling in both post mortem human Parkinson's disease samples and in the 1-methyl-4-phenyl-1, 2, 3, 6-tetrahydropyridine (MPTP) mouse model. In response to MPTP, mice lacking the type-1 IFN receptor (IFNAR1(-/-) ) displayed decreased type-1 IFN signaling, an attenuated pro-inflammatory response and reduced loss of dopaminergic neurons. The neuroprotective potential of targeting the type-1 IFN pathway was confirmed by reduced neuroinflammation and DA cell death in mice treated with a blocking monoclonal IFNAR1 (MAR-1) antibody. The MPTP/MAR-1 treated mice also displayed increased striatal dopamine levels and improved behavioural outcomes compared to their MPTP/IgG controls. These data, implicate for the first time, a deleterious role for the type-1 IFNs as key modulators of the early neuroinflammatory response and therefore the neuronal cell death in Parkinson's disease. GLIA 2016;64:1590-1604.


Assuntos
Neurônios Dopaminérgicos/metabolismo , Interferon Tipo I/genética , Doença de Parkinson/genética , Animais , Morte Celular/genética , Citocinas/metabolismo , Modelos Animais de Doenças , Dopamina/metabolismo , Inflamação/genética , Interferon Tipo I/metabolismo , Camundongos Endogâmicos C57BL , Camundongos Knockout , Microglia/metabolismo , Doença de Parkinson/patologia , Substância Negra/patologia
4.
J Neuroinflammation ; 12: 110, 2015 May 30.
Artigo em Inglês | MEDLINE | ID: mdl-26025142

RESUMO

UNLABELLED: During inflammation, the kynurenine pathway (KP) metabolises the essential amino acid tryptophan (TRP) potentially contributing to excitotoxicity via the release of quinolinic acid (QUIN) and 3-hydroxykynurenine (3HK). Despite the importance of excitotoxicity in the development of secondary brain damage, investigations on the KP in TBI are scarce. In this study, we comprehensively characterised changes in KP activation by measuring numerous metabolites in cerebrospinal fluid (CSF) from TBI patients and assessing the expression of key KP enzymes in brain tissue from TBI victims. Acute QUIN levels were further correlated with outcome scores to explore its prognostic value in TBI recovery. METHODS: Twenty-eight patients with severe TBI (GCS ≤ 8, three patients had initial GCS = 9-10, but rapidly deteriorated to ≤8) were recruited. CSF was collected from admission to day 5 post-injury. TRP, kynurenine (KYN), kynurenic acid (KYNA), QUIN, anthranilic acid (AA) and 3-hydroxyanthranilic acid (3HAA) were measured in CSF. The Glasgow Outcome Scale Extended (GOSE) score was assessed at 6 months post-TBI. Post-mortem brains were obtained from the Australian Neurotrauma Tissue and Fluid Bank and used in qPCR for quantitating expression of KP enzymes (indoleamine 2,3-dioxygenase-1 (IDO1), kynurenase (KYNase), kynurenine amino transferase-II (KAT-II), kynurenine 3-monooxygenase (KMO), 3-hydroxyanthranilic acid oxygenase (3HAO) and quinolinic acid phosphoribosyl transferase (QPRTase) and IDO1 immunohistochemistry. RESULTS: In CSF, KYN, KYNA and QUIN were elevated whereas TRP, AA and 3HAA remained unchanged. The ratios of QUIN:KYN, QUIN:KYNA, KYNA:KYN and 3HAA:AA revealed that QUIN levels were significantly higher than KYN and KYNA, supporting increased neurotoxicity. Amplified IDO1 and KYNase mRNA expression was demonstrated on post-mortem brains, and enhanced IDO1 protein coincided with overt tissue damage. QUIN levels in CSF were significantly higher in patients with unfavourable outcome and inversely correlated with GOSE scores. CONCLUSION: TBI induced a striking activation of the KP pathway with sustained increase of QUIN. The exceeding production of QUIN together with increased IDO1 activation and mRNA expression in brain-injured areas suggests that TBI selectively induces a robust stimulation of the neurotoxic branch of the KP pathway. QUIN's detrimental roles are supported by its association to adverse outcome potentially becoming an early prognostic factor post-TBI.


Assuntos
Lesões Encefálicas/diagnóstico , Lesões Encefálicas/metabolismo , Cinurenina/fisiologia , Neurotoxinas/líquido cefalorraquidiano , Ácido Quinolínico/líquido cefalorraquidiano , Transdução de Sinais/fisiologia , Adolescente , Adulto , Idoso , Biomarcadores/metabolismo , Encéfalo/metabolismo , Lesões Encefálicas/fisiopatologia , Estudos de Casos e Controles , Feminino , Escala de Resultado de Glasgow , Humanos , Indolamina-Pirrol 2,3,-Dioxigenase/metabolismo , Masculino , Pessoa de Meia-Idade , Prognóstico , RNA Mensageiro/metabolismo , Triptofano/sangue , Adulto Jovem
5.
Cell Tissue Res ; 349(2): 565-76, 2012 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-22688956

RESUMO

Recent investigation of the intestine following ischemia and reperfusion (I/R) has revealed that nitric oxide synthase (NOS) neurons are more strongly affected than other neuron types. This implies that NO originating from NOS neurons contributes to neuronal damage. However, there is also evidence of the neuroprotective effects of NO. In this study, we compared the effects of I/R on the intestines of neuronal NOS knockout (nNOS(-/-)) mice and wild-type mice. I/R caused histological damage to the mucosa and muscle and infiltration of neutrophils into the external muscle layers. Damage to the mucosa and muscle was more severe and greater infiltration by neutrophils occurred in the first 24 h in nNOS(-/-) mice. Immunohistochemistry for the contractile protein, α-smooth muscle actin, was used to evaluate muscle damage. Smooth muscle actin occurred in the majority of smooth muscle cells in the external musculature of normal mice but was absent from most cells and was reduced in the cytoplasm of other cells following I/R. The loss was greater in nNOS(-/-) mice. Basal contractile activity of the longitudinal muscle and contractile responses to nerve stimulation or a muscarinic agonist were reduced in regions subjected to I/R and the effects were greater in nNOS(-/-) mice. Reductions in responsiveness also occurred in regions of operated mice not subjected to I/R. This is attributed to post-operative ileus that is not significantly affected by knockout of nNOS. The results indicate that deleterious effects are greater in regions subjected to I/R in mice lacking nNOS compared with normal mice, implying that NO produced by nNOS has protective effects that outweigh any damaging effect of this free radical produced by enteric neurons.


Assuntos
Intestino Delgado/enzimologia , Intestino Delgado/patologia , Óxido Nítrico Sintase Tipo I/genética , Traumatismo por Reperfusão/enzimologia , Traumatismo por Reperfusão/patologia , Actinas/análise , Animais , Feminino , Deleção de Genes , Intestino Delgado/metabolismo , Intestino Delgado/fisiopatologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Contração Muscular , Músculo Liso/enzimologia , Músculo Liso/metabolismo , Músculo Liso/fisiopatologia , Óxido Nítrico/metabolismo , Óxido Nítrico Sintase Tipo I/metabolismo , Traumatismo por Reperfusão/genética , Traumatismo por Reperfusão/fisiopatologia
6.
Cell Mol Neurobiol ; 31(4): 569-77, 2011 May.
Artigo em Inglês | MEDLINE | ID: mdl-21234797

RESUMO

Lysophosphatidic acid (LPA) is involved in physiological and pathological states, including in neural development and inflammation. We assessed the expression pattern of the LPA receptors 1-3 and of LPA-producing enzyme autotaxin in post-mortem human brain tissue, both in normal individuals and in individuals who died following traumatic brain injury. We found that LPA receptors and autotaxin are weakly expressed in the normal control adult brain. Quantitative PCR for the LPA receptors and autotaxin mRNA showed an increase of LPAR(2) and a decrease of autotaxin mRNA expression in the cortex following brain injury. Immunohistochemical analysis showed that LPAR(1) colocalized with astrocytes and that LPAR(2) is present on the ependymal cells lining the lateral ventricle in the brain samples from individuals who died following severe head injury. This work shows for the first time that key components of the LPA pathway are modulated following TBI in humans.


Assuntos
Lesões Encefálicas/genética , Lesões Encefálicas/patologia , Encéfalo/metabolismo , Encéfalo/patologia , Receptores de Ácidos Lisofosfatídicos/genética , Adolescente , Adulto , Idoso , Especificidade de Anticorpos/imunologia , Astrócitos/metabolismo , Astrócitos/patologia , Epêndima/metabolismo , Epêndima/patologia , Feminino , Regulação da Expressão Gênica , Humanos , Masculino , Pessoa de Meia-Idade , Complexos Multienzimáticos/genética , Complexos Multienzimáticos/metabolismo , Fosfodiesterase I/genética , Fosfodiesterase I/metabolismo , Diester Fosfórico Hidrolases , Reação em Cadeia da Polimerase , Pirofosfatases/genética , Pirofosfatases/metabolismo , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Receptores de Ácidos Lisofosfatídicos/metabolismo , Reprodutibilidade dos Testes , Adulto Jovem
7.
J Neuroinflammation ; 7: 67, 2010 Oct 14.
Artigo em Inglês | MEDLINE | ID: mdl-20942978

RESUMO

BACKGROUND: The chemokine CCL2 (also known as monocyte chemoattractant protein-1, or MCP-1) is upregulated in patients and rodent models of traumatic brain injury (TBI), contributing to post-traumatic neuroinflammation and degeneration by directing the infiltration of blood-derived macrophages into the injured brain. Our laboratory has previously reported that Ccl2-/- mice show reduced macrophage accumulation and tissue damage, corresponding to improved motor recovery, following experimental TBI. Surprisingly, Ccl2-deficient mice also exhibited delayed but exacerbated secretion of key proinflammatory cytokines in the injured cortex. Thus we sought to further characterise CCL2's potential ability to modulate immunoactivation of astrocytes in vitro. METHODS: Primary astrocytes were isolated from neonatal wild-type and Ccl2-deficient mice. Established astrocyte cultures were stimulated with various concentrations of lipopolysaccharide (LPS) and interleukin (IL)-1ß for up to 24 hours. Separate experiments involved pre-incubation with mouse recombinant (r)CCL2 prior to IL-1ß stimulation in wild-type cells. Following stimulation, cytokine secretion was measured in culture supernatant by immunoassays, whilst cytokine gene expression was quantified by real-time reverse transcriptase polymerase chain reaction. RESULTS: LPS (0.1-100 µg/ml; 8 h) induced the significantly greater secretion of five key cytokines and chemokines in Ccl2-/- astrocytes compared to wild-type cells. Consistently, IL-6 mRNA levels were 2-fold higher in Ccl2-deficient cells. IL-1ß (10 and 50 ng/ml; 2-24 h) also resulted in exacerbated IL-6 production from Ccl2-/- cultures. Despite this, treatment of wild-type cultures with rCCL2 alone (50-500 ng/ml) did not induce cytokine/chemokine production by astrocytes. However, pre-incubation of wild-type astrocytes with rCCL2 (250 ng/ml, 12 h) prior to stimulation with IL-1ß (10 ng/ml, 8 h) significantly reduced IL-6 protein and gene expression. CONCLUSIONS: Our data indicate that astrocytes are likely responsible for the exacerbated cytokine response seen in vivo post-injury in the absence of CCL2. Furthermore, evidence that CCL2 inhibits cytokine production by astrocytes following IL-1ß stimulation, suggests a novel, immunomodulatory role for this chemokine in acute neuroinflammation. Further investigation is required to determine the physiological relevance of this phenomenon, which may have implications for therapeutics targeting CCL2-mediated leukocyte infiltration following TBI.


Assuntos
Astrócitos/metabolismo , Quimiocina CCL2/metabolismo , Citocinas/biossíntese , Análise de Variância , Animais , Astrócitos/efeitos dos fármacos , Células Cultivadas , Quimiocina CCL2/genética , Citocinas/genética , Lipopolissacarídeos/farmacologia , Camundongos , Camundongos Knockout , Reação em Cadeia da Polimerase Via Transcriptase Reversa
8.
Cell Mol Neurobiol ; 29(5): 621-33, 2009 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-19214740

RESUMO

The transforming growth factor betas (TGFbetas) are context-dependent regulators of neurons in vitro, but their physiological functions in the brain are unclear. Haploinsufficiency of either Tgfbeta1 or Tgfbeta2 leads to age-related deterioration of neurons, but the development of the brain is normal in the full absence of either of these genes. However, some individuals with mis-sense mutations of TGFbeta receptors are mentally retarded, suggesting that the TGFbeta isoforms can compensate for each other during brain development. This possibility was tested by generating mice (NSE x PTR) with neuron-specific expression of a dominant-negative inhibitor of TGFbeta signaling. The NSE x PTR mice with a FVBxC57Bl/6 genetic background were viable and developed normally despite strong neuronal expression of the inhibitor of TGFbeta signaling. Their cerebella were of normal size and contained normal numbers of neurons. When the genetic background of the mice was changed to C57BL/6, the phenotype of the mice became neonatal lethal, with the neonates exhibiting various malformations. The malformations correlated with sites of non-neuronal expression of the transgenes and included facial dysmorphogenesis, incomplete closure of the ventral body wall and absence of intestinal motility. The C57BL/6 Tgfbm1-3 alleles, which modulate the phenotype of Tgfbeta1(-/-) mice, were not major determinants of the NSE x PTR phenotype. The data suggest that the development of the cerebellum is insensitive to the level of TGFbeta signaling, although this may be dependent on the genetic background.


Assuntos
Cerebelo/embriologia , Embrião de Mamíferos/anormalidades , Face/anormalidades , Transdução de Sinais , Fator de Crescimento Transformador beta/metabolismo , Abdome/patologia , Animais , Cruzamentos Genéticos , Doxiciclina/farmacologia , Perda do Embrião/patologia , Embrião de Mamíferos/patologia , Face/patologia , Feminino , Regulação da Expressão Gênica no Desenvolvimento , Masculino , Camundongos , Camundongos Endogâmicos , Fenótipo , Transdução de Sinais/efeitos dos fármacos , Análise de Sobrevida , Transgenes
9.
Curr Opin Genet Dev ; 12(3): 294-8, 2002 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-12076672

RESUMO

Spinal muscular atrophy (SMA) is a common recessive autosomal disorder characterized by degeneration of motor neurons of the spinal cord. SMA is caused by mutations of the survival of motor neuron gene that encodes a multifunctional protein, and mouse models have been generated. These advances represent starting points towards an understanding of the pathophysiology of this disease and the design of therapeutic strategies in SMA.


Assuntos
Atrofia Muscular Espinal/genética , Proteínas do Tecido Nervoso/genética , Animais , Proteína de Ligação ao Elemento de Resposta ao AMP Cíclico , Modelos Animais de Doenças , Conversão Gênica , Deleção de Genes , Humanos , Camundongos , Atrofia Muscular Espinal/etiologia , Atrofia Muscular Espinal/terapia , Proteínas do Tecido Nervoso/fisiologia , Fenótipo , Proteínas de Ligação a RNA , Proteínas do Complexo SMN
10.
J Neurosurg ; : 1-7, 2018 May 04.
Artigo em Inglês | MEDLINE | ID: mdl-29726784

RESUMO

OBJECTIVEDelayed ischemic neurological deficit (DIND) is a leading cause of mortality and morbidity after aneurysmal subarachnoid hemorrhage (aSAH). Arginine vasopressin (AVP) is a hormone released by the posterior pituitary. It is known to cause cerebral vasoconstriction and has been implicated in hyponatremia secondary to the syndrome of inappropriate antidiuretic hormone secretion. Direct measurement of AVP is limited by its short half-life. Copeptin, a cleavage product of the AVP precursor protein, was therefore used as a surrogate marker for AVP. This study aimed to investigate the temporal relationship between changes in copeptin concentrations and episodes of DIND and hyponatremia.METHODSCopeptin concentrations in cerebrospinal fluid were quantified using enzyme-linked immunosorbent assay in 19 patients: 10 patients with DIND, 6 patients without DIND (no-DIND), and 3 controls.RESULTSCopeptin concentrations were higher in DIND and no-DIND patients than in controls. In hyponatremic DIND patients, copeptin concentrations were higher compared with hyponatremic no-DIND patients. DIND was associated with a combination of decreasing sodium levels and increasing copeptin concentrations.CONCLUSIONSIncreased AVP may be the unifying factor explaining the co-occurrence of hyponatremia and DIND. Future studies are indicated to investigate this relationship and the therapeutic utility of AVP antagonists in the clinical setting.

11.
Biochim Biophys Acta ; 1762(10): 890-7, 2006 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-16935476

RESUMO

The neuronal ceroid lipofuscinoses (NCLs, Batten disease) are recessively inherited neurodegenerative disorders that affect humans and other animals, characterised by brain atrophy and the accumulation of lysosome derived fluorescent storage bodies in neurons and most other cells. Common clinical signs include blindness, ataxia, dementia, seizures and premature death. The associated genes for six different human forms have been identified (CLN1, CLN2, CLN3, CLN5, CLN6 and CLN8), and three other human forms suggested (CLNs 4, 7 and 9). A form of NCL in Australian Devon cattle is caused by a single base duplication (c.662dupG) in bovine CLN5. This mutation causes a frame-shift and premature termination (p.Arg221GlyfsX6) which is predicted to result in a severely truncated protein, analogous to disease causing mutations in human Finnish late infantile variant NCL (CLN5), and a simple genetic diagnostic test has been developed. The symptoms and disease course in cattle also matches CLN5. Only one initiation site was found in the bovine gene, equivalent to the third of four possible initiation sites in the human gene. As cattle are anatomically and physiologically similar to humans with a human-like central nervous system and easy to maintain and breed, they provide a valuable alternative model for CLN5 studies.


Assuntos
Doenças dos Bovinos/genética , Bovinos/anormalidades , Mutação da Fase de Leitura , Proteínas de Membrana/genética , Lipofuscinoses Ceroides Neuronais/genética , Lipofuscinoses Ceroides Neuronais/veterinária , Sequência de Aminoácidos , Animais , Cromossomos Artificiais Bacterianos , Predisposição Genética para Doença , Dados de Sequência Molecular , Homologia de Sequência de Aminoácidos , Tripeptidil-Peptidase 1
12.
Biochim Biophys Acta ; 1762(10): 898-905, 2006 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-17046213

RESUMO

The neuronal ceroid lipofuscinoses (NCLs, Batten disease) are a group of fatal recessively inherited neurodegenerative diseases of humans and animals characterised by common clinical signs and pathology. These include blindness, ataxia, dementia, behavioural changes, seizures, brain and retinal atrophy and accumulation of fluorescent lysosome derived organelles in most cells. A number of different variants have been suggested and seven different causative genes identified in humans (CLN1, CLN2, CLN3, CLN5, CLN6, CLN8 and CTSD). Animal models have played a central role in the investigation of this group of diseases and are extremely valuable for developing a better understanding of the disease mechanisms and possible therapeutic approaches. Ovine models include flocks of affected New Zealand South Hampshires and Borderdales and Australian Merinos. The ovine CLN6 gene has been sequenced in a representative selection of these sheep. These investigations unveiled the mutation responsible for the disease in Merino sheep (c.184C>T; p.Arg62Cys) and three common ovine allelic variants (c.56A>G, c.822G>A and c.933_934insCT). Linkage analysis established that CLN6 is the gene most likely to cause NCL in affected South Hampshire sheep, which do not have the c.184C>T mutation but show reduced expression of CLN6 mRNA in a range of tissues as determined by real-time PCR. Lack of linkage precludes CLN6 as a candidate for NCL in Borderdale sheep.


Assuntos
Proteínas de Membrana/genética , Mutação de Sentido Incorreto , Lipofuscinoses Ceroides Neuronais/genética , Lipofuscinoses Ceroides Neuronais/veterinária , Doenças dos Ovinos/genética , Sequência de Aminoácidos , Animais , Ligação Genética , Predisposição Genética para Doença , Dados de Sequência Molecular , RNA Mensageiro/metabolismo , Homologia de Sequência de Aminoácidos , Ovinos , Tripeptidil-Peptidase 1
13.
PLoS One ; 12(11): e0188305, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-29176798

RESUMO

The atypical chemokine receptor ACKR2 promotes resolution of acute inflammation by operating as a scavenger receptor for inflammatory CC chemokines in several experimental models of inflammatory disorders, however its role in the brain remains unclear. Based on our previous reports of increased expression of inflammatory chemokines and their corresponding receptors following traumatic brain injury (TBI), we hypothesised that ACKR2 modulates neuroinflammation following brain trauma and that its deletion exacerbates cellular inflammation and chemokine production. We demonstrate increased CCL2 and ACKR2 mRNA expression in post-mortem human brain, whereby ACKR2 mRNA levels correlated with later times post-TBI. This data is consistent with the transient upregulation of ACKR2 observed in mouse brain after closed head injury (CHI). As compared to WT animals, ACKR2-/- mice showed a higher mortality rate after CHI, while the neurological outcome in surviving mice was similar. At day 1 post-injury, ACKR2-/- mice displayed aggravated lesion volume and no differences in CCL2 expression and macrophage recruitment relative to WT mice. Reciprocal regulation of ACKR2 and CCL2 expression was explored in cultured astrocytes, which are recognized as the major source of CCL2 and also express ACKR2. ACKR2 mRNA increased as early as 2 hours after an inflammatory challenge in WT astrocytes. As expected, CCL2 expression also dramatically increased at 4 hours in WT astrocytes but was significantly lower in ACKR2-/- astrocytes, possibly indicating a co-regulation of CCL2 and ACKR2 in these cells. Conversely, in vivo, CCL2 mRNA/protein levels were increased similarly in ACKR2-/- and WT brains at 4 and 12 hours after CHI, in line with the lack of differences in cerebral macrophage recruitment and neurological recovery. In conclusion, ACKR2 is induced after TBI and has a significant impact on mortality and lesion development acutely following CHI, while its role in chemokine expression, macrophage activation, brain pathology, and neurological recovery at later time-points is minor. Concordant to evidence in multiple sclerosis experimental models, our data corroborate a distinct role for ACKR2 in cerebral inflammatory processes compared to its reported functions in peripheral tissues.


Assuntos
Lesões Encefálicas Traumáticas/metabolismo , Lesões Encefálicas Traumáticas/mortalidade , Receptores de Quimiocinas/metabolismo , Animais , Astrócitos/metabolismo , Astrócitos/patologia , Osso e Ossos/patologia , Encéfalo/metabolismo , Encéfalo/patologia , Encéfalo/fisiopatologia , Lesões Encefálicas Traumáticas/genética , Lesões Encefálicas Traumáticas/fisiopatologia , Células Cultivadas , Quimiocina CCL2/genética , Quimiocina CCL2/metabolismo , Deleção de Genes , Humanos , Inflamação/patologia , Macrófagos/metabolismo , Macrófagos/patologia , Masculino , Camundongos Endogâmicos C57BL , Mortalidade , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Receptores de Quimiocinas/genética , Recuperação de Função Fisiológica , Regulação para Cima/genética
14.
ACS Chem Neurosci ; 8(4): 731-736, 2017 04 19.
Artigo em Inglês | MEDLINE | ID: mdl-28029772

RESUMO

Ferroxidase activity has been reported to be altered in various biological fluids in neurodegenerative disease, but the sources contributing to the altered activity are uncertain. Here we assay fractions of serum and cerebrospinal fluid with a newly validated triplex ferroxidase assay. Our data indicate that while ceruloplasmin, a multicopper ferroxidase, is the predominant source of serum activity, activity in CSF predominantly derives from a <10 kDa component, specifically from polyanions such as citrate and phosphate. We confirm that in human biological samples, ceruloplasmin activity in serum is decreased in Alzheimer's disease, but in CSF a reduction of activity in Alzheimer's disease originates from the polyanion component.


Assuntos
Doença de Alzheimer/metabolismo , Líquido Cefalorraquidiano/enzimologia , Ceruloplasmina/metabolismo , Ferro/metabolismo , Soro/enzimologia , Ceruloplasmina/análise , Humanos , Oxirredução
15.
Neurochem Int ; 96: 62-8, 2016 06.
Artigo em Inglês | MEDLINE | ID: mdl-26930584

RESUMO

Autophagy is a homeostatic process for recycling proteins and organelles that is increasingly being proposed as a therapeutic target for acute and chronic neurodegenerative diseases, including stroke. Confirmation that autophagy is present in the human brain after stroke is imperative before prospective therapies can begin the translational process into clinical trials. Our current study using human post-mortem tissue observed an increase in staining in microtubule-associated protein 1 light chain 3 (LC3), sequestosome 1 (SQSTM1; also known as p62) and the increased appearance of autophagic vesicles after stroke. These data confirm that alterations in autophagy take place in the human brain after stroke and suggest that targeting autophagic processes after stroke may have clinical significance.


Assuntos
Autofagia/fisiologia , Proteína Beclina-1/biossíntese , Encéfalo/metabolismo , Proteínas Associadas aos Microtúbulos/biossíntese , Proteína Sequestossoma-1/biossíntese , Acidente Vascular Cerebral/metabolismo , Idoso , Idoso de 80 Anos ou mais , Proteína Beclina-1/análise , Encéfalo/patologia , Química Encefálica/fisiologia , Feminino , Humanos , Masculino , Proteínas Associadas aos Microtúbulos/análise , Proteína Sequestossoma-1/análise , Acidente Vascular Cerebral/patologia
16.
eNeuro ; 3(1)2016.
Artigo em Inglês | MEDLINE | ID: mdl-27022620

RESUMO

Type-1 interferons (IFNs) are pleiotropic cytokines that signal through the type-1 IFN receptor (IFNAR1). Recent literature has implicated the type-1 IFNs in disorders of the CNS. In this study, we have investigated the role of type-1 IFNs in neuroinflammation following traumatic brain injury (TBI). Using a controlled cortical impact model, TBI was induced in 8- to 10-week-old male C57BL/6J WT and IFNAR1(-/-) mice and brains were excised to study infarct volume, inflammatory mediator release via quantitative PCR analysis and immune cell profile via immunohistochemistry. IFNAR1(-/-) mice displayed smaller infarcts compared with WT mice after TBI. IFNAR1(-/-) mice exhibited an altered anti-inflammatory environment compared with WT mice, with significantly reduced levels of the proinflammatory mediators TNFα, IL-1ß and IL-6, an up-regulation of the anti-inflammatory mediator IL-10 and an increased activation of resident and peripheral immune cells after TBI. WT mice injected intravenously with an anti-IFNAR1 blocking monoclonal antibody (MAR1) 1 h before, 30 min after or 30 min and 2 d after TBI displayed significantly improved histological and behavioral outcome. Bone marrow chimeras demonstrated that the hematopoietic cells are a peripheral source of type-1 IFNs that drives neuroinflammation and a worsened TBI outcome. Type-1 IFN mRNA levels were confirmed to be significantly altered in human postmortem TBI brains. Together, these data demonstrate that type-1 IFN signaling is a critical pathway in the progression of neuroinflammation and presents a viable therapeutic target for the treatment of TBI.


Assuntos
Lesões Encefálicas/metabolismo , Encefalite/metabolismo , Células-Tronco Hematopoéticas/metabolismo , Interferon Tipo I/metabolismo , Receptor de Interferon alfa e beta/metabolismo , Animais , Anticorpos Monoclonais/administração & dosagem , Astrócitos/metabolismo , Encéfalo/metabolismo , Encéfalo/patologia , Lesões Encefálicas/complicações , Lesões Encefálicas/patologia , Encefalite/etiologia , Humanos , Mediadores da Inflamação/metabolismo , Macrófagos/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Microglia/metabolismo , RNA Mensageiro/metabolismo , Receptor de Interferon alfa e beta/antagonistas & inibidores , Receptor de Interferon alfa e beta/genética , Receptor de Interferon alfa e beta/imunologia , Transdução de Sinais
17.
J Appl Physiol (1985) ; 118(9): 1113-21, 2015 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-25749441

RESUMO

Inhibition of nitric oxide synthase (NOS) significantly attenuates the increase in skeletal muscle glucose uptake during contraction/exercise, and a greater attenuation is observed in individuals with Type 2 diabetes compared with healthy individuals. Therefore, NO appears to play an important role in mediating muscle glucose uptake during contraction. In this study, we investigated the involvement of neuronal NOSµ (nNOSµ), the main NOS isoform activated during contraction, on skeletal muscle glucose uptake during ex vivo contraction. Extensor digitorum longus muscles were isolated from nNOSµ(-/-) and nNOSµ(+/+) mice. Muscles were contracted ex vivo in a temperature-controlled (30°C) organ bath with or without the presence of the NOS inhibitor N(G)-monomethyl-l-arginine (L-NMMA) and the NOS substrate L-arginine. Glucose uptake was determined by radioactive tracers. Skeletal muscle glucose uptake increased approximately fourfold during contraction in muscles from both nNOSµ(-/-) and nNOSµ(+/+) mice. L-NMMA significantly attenuated the increase in muscle glucose uptake during contraction in both genotypes. This attenuation was reversed by L-arginine, suggesting that L-NMMA attenuated the increase in muscle glucose uptake during contraction by inhibiting NOS and not via a nonspecific effect of the inhibitor. Low levels of NOS activity (~4%) were detected in muscles from nNOSµ(-/-) mice, and there was no evidence of compensation from other NOS isoform or AMP-activated protein kinase which is also involved in mediating muscle glucose uptake during contraction. These results indicate that NO regulates skeletal muscle glucose uptake during ex vivo contraction independently of nNOSµ.


Assuntos
Glucose/metabolismo , Contração Muscular/fisiologia , Músculo Esquelético/metabolismo , Músculo Esquelético/fisiologia , Óxido Nítrico Sintase Tipo I/metabolismo , Proteínas Quinases Ativadas por AMP/metabolismo , Animais , Arginina/metabolismo , Transporte Biológico/efeitos dos fármacos , Transporte Biológico/fisiologia , Inibidores Enzimáticos/farmacologia , Feminino , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Contração Muscular/efeitos dos fármacos , Músculo Esquelético/efeitos dos fármacos , Óxido Nítrico/metabolismo , Condicionamento Físico Animal/fisiologia , ômega-N-Metilarginina/farmacologia
18.
Metallomics ; 7(1): 66-70, 2015 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-25424382

RESUMO

Traumatic brain injury (TBI) is the most common cause of death and disability in young adults, yet the molecular mechanisms that follow TBI are poorly understood. We previously reported a perturbation in iron (Fe) levels following TBI. Here we report that the distribution of cobalt (Co) is modulated in post-mortem human brain following injury. We also investigated how the distribution of other biologically relevant elements changes in TBI. Cobalt is increased due to TBI while copper (Cu), magnesium (Mg), manganese (Mn), phosphorus (P), potassium (K), rubidium (Rb), selenium (Se) and zinc (Zn) remain unchanged. The elevated Co has important implications for positron emission tomography neuroimaging. This is the first demonstration of the accumulation of Co in injured tissue explaining the previous utility of (55)Co-PET imaging in TBI.


Assuntos
Química Encefálica , Lesões Encefálicas/metabolismo , Cobalto/análise , Cobalto/metabolismo , Adolescente , Adulto , Idoso , Autopsia , Estudos de Coortes , Feminino , Humanos , Masculino , Metais Pesados/análise , Metais Pesados/metabolismo , Pessoa de Meia-Idade , Adulto Jovem
19.
Semin Pediatr Neurol ; 9(2): 145-50, 2002 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-12138998

RESUMO

Spinal muscular atrophies (SMA) are characterized by degeneration of lower motor neurons associated with muscle paralysis and atrophy. Childhood SMA is a common recessive autosomal disorder and represents one of the most common genetic causes of death in childhood. The pathophysiology remains unknown, and no curative treatment is available so far. The last 10 years have seen major advances in the field of SMA, which are starting points towards understanding the SMA pathogenesis and developing therapeutic strategies for this devastating neurodegenerative disease.


Assuntos
Atrofia Muscular Espinal/genética , Cromossomos Humanos Par 5/genética , Proteína de Ligação ao Elemento de Resposta ao AMP Cíclico , Deleção de Genes , Humanos , Músculo Esquelético/patologia , Atrofia Muscular Espinal/patologia , Proteínas do Tecido Nervoso/genética , Mutação Puntual/genética , Proteínas de Ligação a RNA , Proteínas do Complexo SMN
20.
Front Neurosci ; 8: 99, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24847201

RESUMO

A common characteristic of axonopathy is the abnormal accumulation of cytoskeletal proteins. We recently reported that streptozotocin (STZ)-induced type 1 diabetes produced a change in the morphology of sympathetic nerve fibers supplying rat plantar metatarsal arteries (PMAs). Here we investigated whether these morphological changes are associated with axonal accumulation of the type III intermediate filament peripherin and the microtubule protein ß-tubulin III, as both are implicated in axonal remodeling. PMAs from hyperglycemic STZ-treated rats receiving a low dose of insulin (STZ-LI) were compared with those from normoglycemic STZ-treated rats receiving a high dose of insulin (STZ-HI) and vehicle-treated controls. Western blotting revealed an increase in protein expression level for peripherin in PMAs from STZ-LI rats but no change in that for ß-tubulin III. In addition, there was an increase in the number of peripherin immunoreactive nerve fibers in the perivascular nerve plexus of PMAs from STZ-LI rats. Co-labeling for peripherin and neuropeptide Y (a marker for sympathetic axons) revealed that peripherin immunoreactivity increased in sympathetic axons. None of these changes were detected in PMAs from STZ-HI rats, indicating that increased peripherin in sympathetic axons of STZ-LI rats is likely due to hyperglycemia and provides a marker of diabetes-induced nerve damage.

SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA